Your browser doesn't support javascript.
loading
Fc-null anti-PD-1 monoclonal antibodies deliver optimal checkpoint blockade in diverse immune environments.
Moreno-Vicente, Julia; Willoughby, Jane E; Taylor, Martin C; Booth, Steven G; English, Vikki L; Williams, Emily L; Penfold, Christine A; Mockridge, C Ian; Inzhelevskaya, Tatyana; Kim, Jinny; Chan, H T Claude; Cragg, Mark S; Gray, Juliet C; Beers, Stephen A.
Afiliação
  • Moreno-Vicente J; Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, University of Southampton, Southampton, UK.
  • Willoughby JE; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
  • Taylor MC; Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, University of Southampton, Southampton, UK.
  • Booth SG; Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, University of Southampton, Southampton, UK.
  • English VL; Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, University of Southampton, Southampton, UK.
  • Williams EL; Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, University of Southampton, Southampton, UK.
  • Penfold CA; Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, University of Southampton, Southampton, UK.
  • Mockridge CI; Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, University of Southampton, Southampton, UK.
  • Inzhelevskaya T; Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, University of Southampton, Southampton, UK.
  • Kim J; Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, University of Southampton, Southampton, UK.
  • Chan HTC; Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, University of Southampton, Southampton, UK.
  • Cragg MS; Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, University of Southampton, Southampton, UK.
  • Gray JC; Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, University of Southampton, Southampton, UK.
  • Beers SA; Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, University of Southampton, Southampton, UK.
J Immunother Cancer ; 10(1)2022 01.
Article em En | MEDLINE | ID: mdl-35017153
ABSTRACT

BACKGROUND:

Despite extensive clinical use, the mechanisms that lead to therapeutic resistance to anti-programmed cell-death (PD)-1 monoclonal antibodies (mAbs) remain elusive. Here, we sought to determine how interactions between the Fc region of anti-PD-1 mAbs and Fcγ receptors (FcγRs) affect therapeutic activity and how these are impacted by the immune environment.

METHODS:

Mouse and human anti-PD-1 mAbs with different Fc binding profiles were generated and characterized in vitro. The ability of these mAbs to elicit T-cell responses in vivo was first assessed in a vaccination setting using the model antigen ovalbumin. The antitumor activity of anti-PD-1 mAbs was investigated in the context of immune 'hot' MC38 versus 'cold' neuroblastoma tumor models, and flow cytometry performed to assess immune infiltration.

RESULTS:

Engagement of activating FcγRs by anti-PD-1 mAbs led to depletion of activated CD8 T cells in vitro and in vivo, abrogating therapeutic activity. Importantly, the extent of this Fc-mediated modulation was determined by the surrounding immune environment. Low FcγR-engaging mouse anti-PD-1 isotypes, which are frequently used as surrogates for human mAbs, were unable to expand ovalbumin-reactive CD8 T cells, in contrast to Fc-null mAbs. These results were recapitulated in mice expressing human FcγRs, in which clinically relevant hIgG4 anti-PD-1 led to reduced endogenous expansion of CD8 T cells compared with its engineered Fc-null counterpart. In the context of an immunologically 'hot' tumor however, both low-engaging and Fc-null mAbs induced long-term antitumor immunity in MC38-bearing mice. Finally, a similar anti-PD-1 isotype hierarchy was demonstrated in the less responsive 'cold' 9464D neuroblastoma model, where the most effective mAbs were able to delay tumor growth but could not induce long-term protection.

CONCLUSIONS:

Our data collectively support a critical role for FcFcγR interactions in inhibiting immune responses to both mouse and human anti-PD-1 mAbs, and highlight the context-dependent effect that anti-PD-1 mAb isotypes can have on T-cell responses. We propose that engineering of Fc-null anti-PD-1 mAbs would prevent FcγR-mediated resistance in vivo and allow maximal T-cell stimulation independent of the immunological environment.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Receptor de Morte Celular Programada 1 / Inibidores de Checkpoint Imunológico / Imunoterapia / Anticorpos Monoclonais / Neoplasias Limite: Animals / Humans Idioma: En Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Receptor de Morte Celular Programada 1 / Inibidores de Checkpoint Imunológico / Imunoterapia / Anticorpos Monoclonais / Neoplasias Limite: Animals / Humans Idioma: En Ano de publicação: 2022 Tipo de documento: Article